Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Bioengineering human intestinal mucosal grafts using patient-derived organoids, fibroblasts and scaffolds

Abstract

Tissue engineering is an interdisciplinary field that combines stem cells and matrices to form functional constructs that can be used to repair damaged tissues or regenerate whole organs. Tissue stem cells can be expanded and functionally differentiated to form ‘mini-organs’ resembling native tissue architecture and function. The choice of the scaffold is also pivotal to successful tissue reconstruction. Scaffolds may be broadly classified into synthetic or biological depending upon the purpose of the engineered organ. Bioengineered intestinal grafts represent a potential source of transplantable tissue for patients with intestinal failure, a condition resulting from extensive anatomical and functional loss of small intestine and therefore digestive and absorptive capacity. Prior strategies in intestinal bioengineering have predominantly used either murine or pluripotent cells and synthetic or decellularized rodent scaffolds, thus limiting their translation. Microscale models of human intestinal epithelium on shaped hydrogels and synthetic scaffolds are more physiological, but their regenerative potential is limited by scale. Here we present a protocol for bioengineering human intestinal grafts using patient-derived materials in a bioreactor culture system. This includes the isolation, expansion and biobanking of patient-derived intestinal organoids and fibroblasts, the generation of decellularized human intestinal scaffolds from native human tissue and providing a system for recellularization to form transplantable grafts. The duration of this protocol is 12 weeks, and it can be completed by scientists with prior experience of organoid culture. The resulting engineered mucosal grafts comprise physiological intestinal epithelium, matrix and surrounding niche, offering a valuable tool for both regenerative medicine and the study of human gastrointestinal diseases.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of the protocol steps.
Fig. 2: Derivation and expansion of patient-derived intestinal organoids and fibroblasts.
Fig. 3: Generation of decellularized human intestinal scaffolds.
Fig. 4: Recellularization of the scaffold in static culture.
Fig. 5: Bioreactor setup for dynamic culture.
Fig. 6: Characterization of the human intestinal mucosal grafts.

Similar content being viewed by others

Data availability

The data presented in Anticipated results section were previously published and are available via the original primary publication29 and its supplementary information files.

References

  1. Baulies, A., Angelis, N. & Li, V. S. W. Hallmarks of intestinal stem cells. Development 147, dev182675 (2020).

    Article  CAS  Google Scholar 

  2. Clevers, H. The intestinal crypt, a prototype stem cell compartment. Cell 154, 274–284 (2013).

    Article  CAS  Google Scholar 

  3. Heppert, J. K. et al. Transcriptional programmes underlying cellular identity and microbial responsiveness in the intestinal epithelium. Nat. Rev. Gastroenterol. Hepatol. 18, 7–23 (2021).

    Article  Google Scholar 

  4. Pironi, L. et al. ESPEN endorsed recommendations. Definition and classification of intestinal failure in adults. Clin. Nutr. 34, 171–180 (2015).

    Article  Google Scholar 

  5. Pironi, L. Definitions of intestinal failure and the short bowel syndrome. Best. Pract. Res. Clin. Gastroenterol. 30, 173–185 (2016).

    Article  Google Scholar 

  6. Booth, C. C. The metabolic effects of intestinal resection in man. Postgrad. Med. J. 37, 725–739 (1961).

    Article  CAS  Google Scholar 

  7. Tullie, L., Jones, B.C., De Coppi, P. & Li, V.S.W. Building gut from scratch – progress and update of intestinal tissue engineering. Nat. Rev. Gastroenterol. Hepatol. (2022).

  8. Martinez Rivera, A. & Wales, P. W. Intestinal transplantation in children: current status. Pediatr. Surg. Int. 32, 529–540 (2016).

    Article  Google Scholar 

  9. Sato, T. et al. Single Lgr5 stem cells build crypt–villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  Google Scholar 

  10. Fujii, M. & Sato, T. Somatic cell-derived organoids as prototypes of human epithelial tissues and diseases. Nat. Mater. 20, 156–169 (2021).

    Article  CAS  Google Scholar 

  11. Hofer, M. & Lutolf, M. P. Engineering organoids. Nat. Rev. Mater. 6, 402–420 (2021).

    Article  CAS  Google Scholar 

  12. Giobbe, G. G. et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat. Commun. 10, 5658 (2019).

    Article  Google Scholar 

  13. Quarta, M. et al. Bioengineered constructs combined with exercise enhance stem cell-mediated treatment of volumetric muscle loss. Nat. Commun. 8, 15613 (2017).

    Article  CAS  Google Scholar 

  14. Jank, B. J. et al. Engineered composite tissue as a bioartificial limb graft. Biomaterials 61, 246–256 (2015).

    Article  CAS  Google Scholar 

  15. Mazza, G. et al. Decellularized human liver as a natural 3D-scaffold for liver bioengineering and transplantation. Sci. Rep. 5, 13079 (2015).

    Article  CAS  Google Scholar 

  16. Uygun, B. E. et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nat. Med. 16, 814–820 (2010).

    Article  CAS  Google Scholar 

  17. Zhou, H. et al. Bioengineering human lung grafts on porcine matrix. Ann. Surg. 267, 590–598 (2018).

    Article  Google Scholar 

  18. Ott, H. C. et al. Regeneration and orthotopic transplantation of a bioartificial lung. Nat. Med. 16, 927–933 (2010).

    Article  CAS  Google Scholar 

  19. Urbani, L. et al. Multi-stage bioengineering of a layered oesophagus with in vitro expanded muscle and epithelial adult progenitors. Nat. Commun. 9, 4286 (2018).

    Article  Google Scholar 

  20. Rama, P. et al. Limbal stem-cell therapy and long-term corneal regeneration. N. Engl. J. Med. 363, 147–155 (2010).

    Article  CAS  Google Scholar 

  21. Hirsch, T. et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature 551, 327–332 (2017).

    Article  CAS  Google Scholar 

  22. Howard, D., Buttery, L. D., Shakesheff, K. M. & Roberts, S. J. Tissue engineering: strategies, stem cells and scaffolds. J. Anat. 213, 66–72 (2008).

    Article  CAS  Google Scholar 

  23. Gao, M. et al. Tissue-engineered trachea from a 3D-printed scaffold enhances whole-segment tracheal repair. Sci. Rep. 7, 5246 (2017).

    Article  Google Scholar 

  24. Costello, C. M. et al. Synthetic small intestinal scaffolds for improved studies of intestinal differentiation. Biotechnol. Bioeng. 111, 1222–1232 (2014).

    Article  CAS  Google Scholar 

  25. Crapo, P. M., Gilbert, T. W. & Badylak, S. F. An overview of tissue and whole organ decellularization processes. Biomaterials 32, 3233–3243 (2011).

    Article  CAS  Google Scholar 

  26. McDevitt, C. A., Wildey, G. M. & Cutrone, R. M. Transforming growth factor-beta1 in a sterilized tissue derived from the pig small intestine submucosa. J. Biomed. Mater. Res. A 67, 637–640 (2003).

    Article  Google Scholar 

  27. Voytik-Harbin, S. L., Brightman, A. O., Kraine, M. R., Waisner, B. & Badylak, S. F. Identification of extractable growth factors from small intestinal submucosa. J. Cell. Biochem. 67, 478–491 (1997).

    Article  CAS  Google Scholar 

  28. Hodde, J. P., Record, R. D., Liang, H. A. & Badylak, S. F. Vascular endothelial growth factor in porcine-derived extracellular matrix. Endothelium 8, 11–24 (2001).

    Article  CAS  Google Scholar 

  29. Meran, L. et al. Engineering transplantable jejunal mucosal grafts using patient-derived organoids from children with intestinal failure. Nat. Med. 26, 1593–1601 (2020).

    Article  CAS  Google Scholar 

  30. Shultz, L. D. et al. Subcapsular transplantation of tissue in the kidney. Cold Spring Harb. Protoc. 2014, 737–740 (2014).

    Article  Google Scholar 

  31. Obokata, H., Yamato, M., Tsuneda, S. & Okano, T. Reproducible subcutaneous transplantation of cell sheets into recipient mice. Nat. Protoc. 6, 1053–1059 (2011).

    Article  CAS  Google Scholar 

  32. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  CAS  Google Scholar 

  33. Fujii, M., Matano, M., Nanki, K. & Sato, T. Efficient genetic engineering of human intestinal organoids using electroporation. Nat. Protoc. 10, 1474–1485 (2015).

    Article  CAS  Google Scholar 

  34. Drost, J., Artegiani, B. & Clevers, H. The generation of organoids for studying Wnt signaling. Methods Mol. Biol. 1481, 141–159 (2016).

    Article  CAS  Google Scholar 

  35. Yin, X. et al. Niche-independent high-purity cultures of Lgr5+ intestinal stem cells and their progeny. Nat. Methods 11, 106–112 (2014).

    Article  CAS  Google Scholar 

  36. Vangipuram, M., Ting, D., Kim, S., Diaz, R. & Schule, B. Skin punch biopsy explant culture for derivation of primary human fibroblasts. J. Vis. Exp. e3779 (2013).

  37. Miller, R. C., Hiraoka, T., Enno, M. & Takeichi, N. Recovery from radiation-induced damage in primary cultures of human epithelial thyroid cells. J. Radiat. Res. 26, 346–352 (1985).

    Article  CAS  Google Scholar 

  38. Meezan, E., Hjelle, J. T., Brendel, K. & Carlson, E. C. A simple, versatile, nondisruptive method for the isolation of morphologically and chemically pure basement membranes from several tissues. Life Sci. 17, 1721–1732 (1975).

    Article  CAS  Google Scholar 

  39. Totonelli, G. et al. A rat decellularized small bowel scaffold that preserves villus–crypt architecture for intestinal regeneration. Biomaterials 33, 3401–3410 (2012).

    Article  CAS  Google Scholar 

  40. Urbani, L. et al. Long-term cryopreservation of decellularised oesophagi for tissue engineering clinical application. PLoS ONE 12, e0179341 (2017).

    Article  Google Scholar 

  41. Fragkos, K. C. & Forbes, A. Citrulline as a marker of intestinal function and absorption in clinical settings: A systematic review and meta-analysis. U. Eur. Gastroenterol. J. 6, 181–191 (2018).

    Article  CAS  Google Scholar 

  42. Boyde, T. R. & Rahmatullah, M. Optimization of conditions for the colorimetric determination of citrulline, using diacetyl monoxime. Anal. Biochem. 107, 424–431 (1980).

    Article  CAS  Google Scholar 

  43. Ray, K. Next-generation intestinal organoids. Nat. Rev. Gastroenterol. Hepatol. 17, 649–649 (2020).

    Article  Google Scholar 

  44. Campinoti, S. et al. Reconstitution of a functional human thymus by postnatal stromal progenitor cells and natural whole-organ scaffolds. Nat. Commun. 11, 6372 (2020).

    Article  CAS  Google Scholar 

  45. Kitano, K. et al. Bioengineering of functional human induced pluripotent stem cell-derived intestinal grafts. Nat. Commun. 8, 765 (2017).

    Article  Google Scholar 

  46. Palikuqi, B. et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature 585, 426–432 (2020).

    Article  CAS  Google Scholar 

  47. Progatzky, F. et al. Regulation of intestinal immunity and tissue repair by enteric glia. Nature 599, 125–130 (2021).

    Article  CAS  Google Scholar 

  48. McCann, C. J. et al. Transplantation of enteric nervous system stem cells rescues nitric oxide synthase deficient mouse colon. Nat. Commun. 8, 15937 (2017).

    Article  CAS  Google Scholar 

  49. Choi, R. S. & Vacanti, J. P. Preliminary studies of tissue-engineered intestine using isolated epithelial organoid units on tubular synthetic biodegradable scaffolds. Transplant. Proc. 29, 848–851 (1997).

    Article  CAS  Google Scholar 

  50. Grikscheit, T. C. et al. Tissue-engineered small intestine improves recovery after massive small bowel resection. Ann. Surg. 240, 748–754 (2004).

    Article  Google Scholar 

  51. Spence, J. R. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109 (2011).

    Article  Google Scholar 

  52. Finkbeiner, S. R. et al. Generation of tissue-engineered small intestine using embryonic stem cell-derived human intestinal organoids. Biol. Open 4, 1462–1472 (2015).

    Article  CAS  Google Scholar 

  53. Finkbeiner, S.R. et al. Transcriptome-wide analysis reveals hallmarks of human intestine development and maturation in vitro and in vivo. Stem Cell Rep. (2015).

  54. Sugimoto, S. et al. An organoid-based organ-repurposing approach to treat short bowel syndrome. Nature 592, 99–104 (2021).

    CAS  Google Scholar 

  55. Cromeens, B. P. et al. Production of tissue-engineered intestine from expanded enteroids. J. Surg. Res. 204, 164–175 (2016).

    Article  CAS  Google Scholar 

  56. Shaffiey, S. A. et al. Intestinal stem cell growth and differentiation on a tubular scaffold with evaluation in small and large animals. Regen. Med. 11, 45–61 (2016).

    Article  CAS  Google Scholar 

  57. Wang, Y. et al. Formation of human colonic crypt array by application of chemical gradients across a shaped epithelial monolayer. Cell Mol. Gastroenterol. Hepatol. 5, 113–130 (2018).

    Article  Google Scholar 

  58. Nikolaev, M. et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 585, 574–578 (2020).

    Article  Google Scholar 

  59. Clevers, H. et al. Tissue-engineering the intestine: the trials before the trials. Cell Stem Cell 24, 855–859 (2019).

    Article  CAS  Google Scholar 

  60. Spencer, A. U. et al. Pediatric short bowel syndrome: redefining predictors of success. Ann. Surg. 242, 403–409 (2005). discussion 409-412.

    Article  Google Scholar 

  61. Wang, X. et al. Cloning and variation of ground state intestinal stem cells. Nature 522, 173–178 (2015).

    Article  CAS  Google Scholar 

  62. Hu, S. et al. Surface modification of poly(dimethylsiloxane) microfluidic devices by ultraviolet polymer grafting. Anal. Chem. 74, 4117–4123 (2002).

    Article  CAS  Google Scholar 

  63. Driehuis, E., Kretzschmar, K. & Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 15, 3380–3409 (2020).

    Article  CAS  Google Scholar 

  64. Jung, P. et al. Isolation and in vitro expansion of human colonic stem cells. Nat. Med. 17, 1225–1227 (2011).

    Article  CAS  Google Scholar 

  65. Gilbert, T. W., Freund, J. M. & Badylak, S. F. Quantification of DNA in biologic scaffold materials. J. Surg. Res. 152, 135–139 (2009).

    Article  CAS  Google Scholar 

  66. Badylak, S. F. & Gilbert, T. W. Immune response to biologic scaffold materials. Semin. Immunol. 20, 109–116 (2008).

    Article  CAS  Google Scholar 

  67. Booth, C. & O’Shea, J. Isolation and culture of intestinal epithelial cells. in Culture of Epithelial Cells (eds. Freshney, R. & Freshney, M.) 303–335 (Wiley-Liss, 2002).

  68. Pleguezuelos-Manzano, C. et al. Establishment and culture of human intestinal organoids derived from adult stem cells. Curr. Protoc. Immunol. 130, e106 (2020).

    Article  CAS  Google Scholar 

  69. Bigaeva, E. et al. Growth factors of stem cell niche extend the life-span of precision-cut intestinal slices in culture: a proof-of-concept study. Toxicol. Vitr. 59, 312–321 (2019).

    Article  CAS  Google Scholar 

  70. Fischer, A. H., Jacobson, K. A., Rose, J. & Zeller, R. Hematoxylin and eosin staining of tissue and cell sections. Cold Spring Harb. Protoc. 2008, pdb prot4986 (2008).

    Article  Google Scholar 

  71. Svensson, B. et al. An amphiphilic form of dipeptidyl peptidase IV from pig small-intestinal brush-border membrane. Purification by immunoadsorbent chromatography and some properties. Eur. J. Biochem. 90, 489–498 (1978).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Brock, from the Research Illustration and Graphics team, the Francis Crick Institute, for contributions to figure illustration and M. Orford for assistance in developing the citrulline assay. We also thank the Francis Crick Institute’s Experimental Histopathology Science Technology Platform for technical support. Selected illustrations in Figs. 1 and 2 were created with BioRender.com. This work was funded by Horizon 2020 grant INTENS 668294 on the project ‘Intestinal Tissue Engineering Solution for Children with Short Bowel Syndrome.’ The laboratory of V.S.W.L. is supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (FC001105), the UK Medical Research Council (FC001105) and the Wellcome Trust (FC001105). P.D.C. is supported by an NIHR Professorship, NIHR UCL BRC-GOSH, the Great Ormond Street Hospital Children’s Charity and the Oak Foundation. L.M. was funded by NIHR UCL BRC-GOSH Crick Clinical Research Training Fellowship and is currently funded by an NIHR Academic Clinical Lectureship. L.T. is funded by NIHR UCL BRC-GOSH Crick Clinical Research Training Fellowship and Horizon 2020 grant INTENS 668294. We are grateful to Gastroenterology, the SNAPS Unit and patients at Great Ormond Street Hospital Children NHS Trust for the intestinal samples and to P. Shi Chia for help with research consents and sample collection.

Author information

Authors and Affiliations

Authors

Contributions

L.M. developed the protocols. L.M. and L.T. performed the experiments and wrote the manuscript. S.E. advised on citrulline assay analyses. P.D.C. and V.S.W.L. edited the manuscript, supervised the study and acquired funding for the study.

Corresponding authors

Correspondence to Paolo De Coppi or Vivian S. W. Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Shinya Sugimot and Shiro Yui for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Meran, L. et al. Nat. Med. 26, 1593–1601 (2020): https://doi.org/10.1038/s41591-020-1024-z

Totonelli, G. et al. Biomaterials 33, 3401–3410 (2012): https://doi.org/10.1016/j.biomaterials.2012.01.012

Giobbe, G. G. et al. Nat. Commun. 10, 5658 (2019): https://doi.org/10.1038/s41467-019-13605-4

Supplementary information

Supplementary Information

Supplementary Methods and Table 1

Reporting Summary

Supplementary Video 1

Bioreactor circuit components setup 1

Supplementary Video 2

Bioreactor circuit components setup 2

Supplementary Video 3

Bioreactor circuit components setup 3

Supplementary Video 4

Bioreactor circuit components setup 4

Supplementary Video 5

Preparation of scaffold

Supplementary Video 6

Mounting of scaffold

Supplementary Video 7

Injection of fibroblasts into scaffold

Supplementary Video 8

Dynamic culture circuit setup 1

Supplementary Video 9

Dynamic culture circuit setup 2

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Meran, L., Tullie, L., Eaton, S. et al. Bioengineering human intestinal mucosal grafts using patient-derived organoids, fibroblasts and scaffolds. Nat Protoc 18, 108–135 (2023). https://doi.org/10.1038/s41596-022-00751-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00751-1

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research